Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Curr Med Res Opin ; 38(12): 2013-2020, 2022 12.
Article in English | MEDLINE | ID: mdl-35791687

ABSTRACT

BACKGROUND: This study aimed to describe the neurological improvements in a patient with severe long COVID brain dysfunction following perispinal etanercept administration. Perispinal administration of etanercept, a novel method designed to enhance its brain delivery via carriage in the cerebrospinal venous system, has previously been shown to reduce chronic neurological dysfunction after stroke. Etanercept is a recombinant biologic that is capable of ameliorating two components of neuroinflammation: microglial activation and the excess bioactivity of tumor necrosis factor (TNF), a proinflammatory cytokine that is a key neuromodulator in the brain. Optimal synaptic and brain network function require physiological levels of TNF. Neuroinflammation, including brain microglial activation and excess central TNF, can be a consequence of stroke or peripheral infection, including infection by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes COVID-19. METHODS: Standardized, validated measures, including the Montreal Cognitive Assessment, Beck Depression Index-II (BDI-II), Fatigue Assessment Scale, Controlled Oral Word Association Test, Trail Making Tests, Timed Finger-to-Nose Test, 20 m Self-Paced Walk Test, 5 Times Sit-to-Stand Test and Grip Strength measured with a Jamar Dynamometer were used to quantitate changes in cognition, depression, fatigue and neurological function after a single 25 mg perispinal etanercept dose in a patient with severe long COVID of 12 months duration. RESULTS: Following perispinal etanercept administration there was immediate neurological improvement. At 24 h, there were remarkable reductions in chronic post-COVID-19 fatigue and depression, and significant measurable improvements in cognition, executive function, phonemic verbal fluency, balance, gait, upper limb coordination and grip strength. Cognition, depression and fatigue were examined at 29 days; each remained substantially improved. CONCLUSION: Perispinal etanercept is a promising treatment for the chronic neurologic dysfunction that may persist after resolution of acute COVID-19, including chronic cognitive dysfunction, fatigue, and depression. These results suggest that long COVID brain neuroinflammation is a potentially reversible pathology and viable treatment target. In view of the increasing unmet medical need, clinical trials of perispinal etanercept for long COVID are urgently necessary. The robust results of the present case suggest that perispinal etanercept clinical trials studying long COVID populations with severe fatigue, depression and cognitive dysfunction may have improved ability to detect a treatment effect. Positron emission tomographic methods that image brain microglial activation and measurements of cerebrospinal fluid proinflammatory cytokines may be useful for patient selection and correlation with treatment effects, as well as provide insight into the underlying pathophysiology.


Subject(s)
COVID-19 Drug Treatment , COVID-19 , Stroke , Humans , Etanercept/therapeutic use , Receptors, Tumor Necrosis Factor/therapeutic use , COVID-19/complications , Immunoglobulin G/therapeutic use , SARS-CoV-2 , Tumor Necrosis Factor-alpha , Stroke/drug therapy , Fatigue , Post-Acute COVID-19 Syndrome
2.
Brain Res ; 1770: 147642, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34474000

ABSTRACT

Disruption of the blood-brain barrier (BBB) is a critical component of traumatic brain injury (TBI) progression. However, further research into the mechanism of BBB disruption and its specific role in TBI pathophysiology is necessary. To help make progress in elucidating TBI affected BBB pathophysiology, we report herein relative gene expression of eleven TBI biomarkers and other factors of neuronal function in human brain microvascular cells (HBMVEC), one of the main cell types in the BBB. Our in-vitro blast TBI model employs a custom acoustic shock tube to deliver injuries of varying intensities to HBMVECs in culture. Each of the investigated genes exhibit a significant change in expression as a response to TBI, which is dependent on both the injury intensity and time following the injury. This data suggests that cell signaling of HBMVECs could be essential to understanding the interaction of the BBB and TBI pathophysiology, warranting future investigation.


Subject(s)
Blast Injuries/metabolism , Blood-Brain Barrier/metabolism , Brain Injuries, Traumatic/metabolism , Brain/metabolism , Endothelial Cells/metabolism , Gene Expression , Biomarkers/metabolism , Blast Injuries/genetics , Blast Injuries/pathology , Blood-Brain Barrier/pathology , Brain/pathology , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/pathology , Endothelial Cells/pathology , Humans
3.
Front Pharmacol ; 12: 817236, 2021.
Article in English | MEDLINE | ID: mdl-35126148

ABSTRACT

Chemotherapy-induced peripheral neurotoxicity is a common dose-limiting side effect of several cancer chemotherapeutic agents, and no effective therapies exist. Here we constructed a systems pharmacology model of intracellular signaling in peripheral neurons to identify novel drug targets for preventing peripheral neuropathy associated with proteasome inhibitors. Model predictions suggested the combinatorial inhibition of TNFα, NMDA receptors, and reactive oxygen species should prevent proteasome inhibitor-induced neuronal apoptosis. Dexanabinol, an inhibitor of all three targets, partially restored bortezomib-induced reduction of proximal action potential amplitude and distal nerve conduction velocity in vitro and prevented bortezomib-induced mechanical allodynia and thermal hyperalgesia in rats, including a partial recovery of intraepidermal nerve fiber density. Dexanabinol failed to restore bortezomib-induced decreases in electrophysiological endpoints in rats, and it did not compromise bortezomib anti-cancer effects in U266 multiple myeloma cells and a murine xenograft model. Owing to its favorable safety profile in humans and preclinical efficacy, dexanabinol might represent a treatment option for bortezomib-induced neuropathic pain.

4.
Front Cell Neurosci ; 14: 233, 2020.
Article in English | MEDLINE | ID: mdl-33005129

ABSTRACT

Schizophrenia (SZ) is a neurodevelopmental genetic disorder in which maternal immune activation (MIA) and increased tumor necrosis factor-α (TNF-α) may contribute. Previous studies using iPSC-derived cerebral organoids and neuronal cells demonstrated developmental malformation and transcriptional dysregulations, including TNF receptors and their signaling genes, common to SZ patients with diverse genetic backgrounds. In the present study, we examined the significance of the common TNF receptor dysregulations by transiently exposing cerebral organoids from embryonic stem cells (ESC) and from representative control and SZ patient iPSCs to TNF. In control iPSC organoids, TNF produced malformations qualitatively similar in, but generally less pronounced than, the malformations of the SZ iPSC-derived organoids. TNF and SZ alone disrupted subcortical rosettes and dispersed proliferating Ki67+ neural progenitor cells (NPC) from the organoid ventricular zone (VZ) into the cortical zone (CZ). In the CZ, the absence of large ramified pan-Neu+ neurons coincided with loss of myelinated neurites despite increased cortical accumulation of O4+ oligodendrocytes. The number of calretinin+ interneurons increased; however, they lacked the preferential parallel orientation to the organoid surface. SZ and SZ+TNF affected fine cortical and subcortical organoid structure by replacing cells with extracellular matrix (ECM)-like fibers The SZ condition increased developmental vulnerability to TNF, leading to more pronounced changes in NPC, pan-Neu+ neurons, and interneurons. Both SZ- and TNF-induced malformations were associated with the loss of nuclear (n)FGFR1 form in the CZ and its upregulation in deep IZ regions, while in earlier studies blocking nFGFR1 reproduced cortical malformations observed in SZ. Computational analysis of ChiPseq and RNAseq datasets shows that nFGFR1 directly targets neurogenic, oligodendrogenic, cell migration, and ECM genes, and that the FGFR1-targeted TNF receptor and signaling genes are overexpressed in SZ NPC. Through these changes, the developing brain with the inherited SZ genome dysregulation may suffer increased vulnerability to TNF and thus, MIA.

5.
Brain Behav Immun ; 82: 93-105, 2019 11.
Article in English | MEDLINE | ID: mdl-31376497

ABSTRACT

Neuropathic pain is chronic pain that follows nerve injury, mediated in the brain by elevated levels of the inflammatory protein tumor necrosis factor-alpha (TNF). We have shown that peripheral nerve injury increases TNF in the hippocampus/pain perception region, which regulates neuropathic pain symptoms. In this study we assessed pain sensation and perception subsequent to specific targeting of brain-TNF (via TNF antibody) administered through a novel subcutaneous perispinal route. Neuropathic pain was induced in Sprague-Dawley rats via chronic constriction injury (CCI), and thermal hyperalgesia was monitored for 10 days post-surgery. On day 8 following CCI and sensory pain behavior testing, rats were randomized to receive perispinal injection of TNF antibody or control IgG isotype antibody. Pain perception was assessed using conditioned place preference (CPP) to the analgesic, amitriptyline. CCI-rats receiving the perispinal injection of TNF antibody had significantly decreased CCI-induced thermal hyperalgesia the following day, and did not form an amitriptyline-induced CPP, whereas CCI-rats receiving perispinal IgG antibody experienced pain alleviation only in conjunction with i.p. amitriptyline and did form an amitriptyline-induced CPP. The specific targeting of brain TNF via perispinal delivery alleviates thermal hyperalgesia and positively influences the affective component of pain. PERSPECTIVE: This study presents a novel route of drug administration to target central TNF for treatment of neuropathic pain. Targeting central TNF through perispinal drug delivery could potentially be a more efficient and sustained method to treat patients with neuropathic pain.


Subject(s)
Neuralgia/drug therapy , Pain Perception/drug effects , Analgesics/administration & dosage , Analgesics/pharmacology , Animals , Brain/metabolism , Chronic Pain/metabolism , Conditioning, Psychological , Hippocampus/drug effects , Hippocampus/metabolism , Hyperalgesia/metabolism , Injections, Intramuscular/methods , Male , Neuralgia/metabolism , Pain Threshold/drug effects , Peripheral Nerve Injuries/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord/metabolism , Tumor Necrosis Factor Inhibitors/administration & dosage , Tumor Necrosis Factor Inhibitors/pharmacology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
6.
7.
Biochem Pharmacol ; 153: 260-268, 2018 07.
Article in English | MEDLINE | ID: mdl-29366977

ABSTRACT

Chronic pain is a large, unmet public health problem. Recent studies have demonstrated the importance of neuroinflammation in the establishment and maintenance of chronic pain. However, pharmacotherapies that reduce neuroinflammation have not been successfully developed to treat chronic pain thus far. Several preclinical studies have established imidazoline I2 receptor (I2R) agonists as novel candidates for chronic pain therapies, and while some I2R ligands appear to modulate neuroinflammation in certain scenarios, whether they exert anti-neuroinflammatory effects in models of chronic pain is unknown. This study examined the effects of the prototypical I2R agonist 2-(2-benzofuranyl)-2-imidazoline hydrochloride (2-BFI) on hypersensitivity and neuroinflammation induced by chronic constriction injury (CCI), a neuropathic pain model in rats. In CCI rats, twice-daily treatment with 10 mg/kg 2-BFI for seven days consistently increased mechanical and thermal nociception thresholds, reduced GFAP and Iba-1 levels in the dorsal horn of the spinal cord, and reduced levels of TNF-α relative to saline treatment. These results were recapitulated in primary mouse cortical astrocyte cultures. Incubation with 2-BFI attenuated GFAP expression and supernatant TNF-α levels in LPS-stimulated cultures. These results suggest that I2R agonists such as 2-BFI may reduce neuroinflammation which may partially account for their antinociceptive effects.


Subject(s)
Benzofurans/therapeutic use , Disease Models, Animal , Hyperalgesia/drug therapy , Imidazoles/therapeutic use , Imidazoline Receptors/agonists , Neuralgia/drug therapy , Spinal Cord Dorsal Horn/drug effects , Animals , Benzofurans/pharmacology , Cells, Cultured , Hyperalgesia/metabolism , Hyperalgesia/pathology , Imidazoles/pharmacology , Imidazoline Receptors/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Neuralgia/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord Dorsal Horn/metabolism , Spinal Cord Dorsal Horn/pathology
8.
Neurosci Biobehav Rev ; 53: 139-59, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25857253

ABSTRACT

Major depression and chronic pain are significant health problems that seriously impact the quality of life of affected individuals. These diseases that individually are difficult to treat often co-exist, thereby compounding the patient's disability and impairment as well as the challenge of successful treatment. The development of efficacious treatments for these comorbid disorders requires a more comprehensive understanding of their linked associations through common neuromodulators, such as tumor necrosis factor-α (TNFα), and various neurotransmitters, as well as common neuroanatomical pathways and structures, including the hippocampal brain region. This review discusses the interaction between depression and chronic pain, emphasizing the fundamental role of the hippocampus in the development and maintenance of both disorders. The focus of this review addresses the hypothesis that hippocampal expressed TNFα serves as a therapeutic target for management of chronic pain and major depressive disorder (MDD).


Subject(s)
Chronic Pain/metabolism , Depressive Disorder, Major/metabolism , Hippocampus/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Chronic Pain/complications , Chronic Pain/physiopathology , Depressive Disorder, Major/complications , Depressive Disorder, Major/physiopathology , Disease Models, Animal , Encephalitis/complications , Encephalitis/metabolism , Humans , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiopathology , Neurons/metabolism , Neurons/physiology , Pain Perception/physiology , Pituitary-Adrenal System/metabolism , Pituitary-Adrenal System/physiopathology , Stress, Psychological/complications , Stress, Psychological/metabolism , Stress, Psychological/physiopathology
9.
Pain ; 156(7): 1320-1333, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25851457

ABSTRACT

Neuropathic pain is a chronic pain syndrome that arises from nerve injury. Current treatments only offer limited relief, clearly indicating the need for more effective therapeutic strategies. Previously, we demonstrated that proinflammatory tumor necrosis factor-alpha (TNF) is a key mediator of neuropathic pain pathogenesis; TNF is elevated at sites of neuronal injury, in the spinal cord, and supraspinally during the initial development of pain. The inhibition of TNF action along pain pathways outside higher brain centers results in transient decreases in pain perception. The objective of this study was to determine whether specific blockade of TNF in the hippocampus, a site of pain integration, could prove efficacious in reducing sciatic nerve chronic constriction injury (CCI)-induced pain behavior. Small inhibitory RNA directed against TNF mRNA was complexed to gold nanorods (GNR-TNF siRNA; TNF nanoplexes) and injected into the contralateral hippocampus of rats 4 days after unilateral CCI. Withdrawal latencies to a noxious thermal stimulus (hyperalgesia) and withdrawal to innocuous forces (allodynia) were recorded up to 10 days and compared with baseline values and sham-operated rats. Thermal hyperalgesia was dramatically decreased in CCI rats receiving hippocampal TNF nanoplexes; and mechanical allodynia was transiently relieved. TNF levels (bioactive protein, TNF immunoreactivity) in hippocampal tissue were decreased. The observation that TNF nanoplex injection into the hippocampus alleviated neuropathic pain-like behavior advances our previous findings that hippocampal TNF levels modulate pain perception. These data provide evidence that targeting TNF in the brain using nanoparticle-protected siRNA may be an effective strategy for treatment of neuropathic pain.


Subject(s)
Hippocampus/metabolism , Nanomedicine/methods , Nanotubes , Nociceptive Pain/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Animals , Constriction , Hippocampus/drug effects , Male , Nociceptive Pain/drug therapy , RNA, Small Interfering/administration & dosage , Rats , Rats, Sprague-Dawley
11.
CNS Drugs ; 28(8): 679-97, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24861337

ABSTRACT

There is increasing recognition of the involvement of the immune signaling molecule, tumor necrosis factor (TNF), in the pathophysiology of stroke and chronic brain dysfunction. TNF plays an important role both in modulating synaptic function and in the pathogenesis of neuropathic pain. Etanercept is a recombinant therapeutic that neutralizes pathologic levels of TNF. Brain imaging has demonstrated chronic intracerebral microglial activation and neuroinflammation following stroke and other forms of acute brain injury. Activated microglia release TNF, which mediates neurotoxicity in the stroke penumbra. Recent observational studies have reported rapid and sustained improvement in chronic post-stroke neurological and cognitive dysfunction following perispinal administration of etanercept. The biological plausibility of these results is supported by independent evidence demonstrating reduction in cognitive dysfunction, neuropathic pain, and microglial activation following the use of etanercept, as well as multiple studies reporting improvement in stroke outcome and cognitive impairment following therapeutic strategies designed to inhibit TNF. The causal association between etanercept treatment and reduction in post-stroke disability satisfy all of the Bradford Hill Criteria: strength of the association; consistency; specificity; temporality; biological gradient; biological plausibility; coherence; experimental evidence; and analogy. Recognition that chronic microglial activation and pathologic TNF concentration are targets that may be therapeutically addressed for years following stroke and other forms of acute brain injury provides an exciting new direction for research and treatment.


Subject(s)
Cognition Disorders/drug therapy , Cognition Disorders/etiology , Immunoglobulin G/administration & dosage , Neuroprotective Agents/administration & dosage , Receptors, Tumor Necrosis Factor/administration & dosage , Stroke/complications , Stroke/drug therapy , Animals , Brain/drug effects , Brain/physiopathology , Cognition Disorders/physiopathology , Etanercept , Humans , Stroke/physiopathology
12.
Clin Drug Investig ; 34(5): 361-6, 2014 May.
Article in English | MEDLINE | ID: mdl-24647830

ABSTRACT

Positron emission tomographic brain imaging and pathological examination have revealed that a chronic, intracerebral neuroinflammatory response lasting for years after a single brain injury may occur in humans. Evidence suggests the immune signaling molecule, tumor necrosis factor (TNF), is centrally involved in this pathology through its modulation of microglial activation, role in synaptic dysfunction, and induction of depressive symptoms and neuropathic pain. Etanercept is a recombinant TNF receptor fusion protein and potent TNF inhibitor that has been found to reduce microglial activation and neuropathic pain in multiple experimental models. We report that a single dose of perispinal etanercept produced an immediate, profound, and sustained improvement in expressive aphasia, speech apraxia, and left hemiparesis in a patient with chronic, intractable, debilitating neurological dysfunction present for more than 3 years after acute brain injury. These results indicate that acute brain injury-induced pathologic levels of TNF may provide a therapeutic target that can be addressed years after injury. Perispinal administration of etanercept is capable of producing immediate relief from brain injury-mediated neurological dysfunction.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Brain Injuries/drug therapy , Immunoglobulin G/administration & dosage , Immunoglobulin G/therapeutic use , Receptors, Tumor Necrosis Factor/administration & dosage , Receptors, Tumor Necrosis Factor/therapeutic use , Recovery of Function/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Brain Injuries/immunology , Brain Injuries/pathology , Brain Injuries/physiopathology , Etanercept , Female , Humans , Immunoglobulin G/pharmacology , Middle Aged , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
13.
Pain ; 153(9): 1871-1882, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22770843

ABSTRACT

The manifestation of chronic, neuropathic pain includes elevated levels of the cytokine tumor necrosis factor-alpha (TNF). Previously, we have shown that the hippocampus, an area of the brain most notable for its role in learning and memory formation, plays a fundamental role in pain sensation. Using an animal model of peripheral neuropathic pain, we have demonstrated that intracerebroventricular infusion of a TNF antibody adjacent to the hippocampus completely alleviated pain. Furthermore, intracerebroventricular infusion of rTNF adjacent to the hippocampus induced pain behavior in naïve animals similar to that expressed during a model of neuropathic pain. These data support our premise that enhanced production of hippocampal-TNF is integral in pain sensation. In the present study, TNF gene expression was induced exclusively in the hippocampus, eliciting increased local bioactive TNF levels, and animals were assessed for pain behaviors. Male Sprague-Dawley rats received stereotaxic injection of gold nanorod (GNR)-complexed cDNA (control or TNF) plasmids (nanoplasmidexes), and pain responses (i.e., thermal hyperalgesia and mechanical allodynia) were measured. Animals receiving hippocampal microinjection of TNF nanoplasmidexes developed thermal hyperalgesia bilaterally. Sensitivity to mechanical stimulation also developed bilaterally in the rat hind paws. In support of these behavioral findings, immunoreactive staining for TNF, bioactive levels of TNF, and levels of TNF mRNA per polymerase chain reaction analysis were assessed in several brain regions and found to be increased only in the hippocampus. These findings indicate that the specific elevation of TNF in the hippocampus is not a consequence of pain, but in fact induces these behaviors/symptoms.


Subject(s)
Hippocampus/metabolism , Hyperalgesia/metabolism , Pain/metabolism , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , DNA, Complementary , Gene Expression , Hot Temperature , Male , Nanotubes , Plasmids , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Touch , Tumor Necrosis Factor-alpha/genetics
14.
Nanomedicine (Lond) ; 6(4): 617-30, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21718174

ABSTRACT

AIM: Gold nanorods (GNRs), cellular imaging nanoprobes, have been used for drug delivery therapy to immunologically privileged regions in the brain. We demonstrate that nanoplexes formed by electrostatic binding between negatively charged RNA and positively charged GNRs, silence the expression of the target housekeeping gene, glyceraldehyde 3-phosphate dehydrogenase (GAPDH) within the CA1 hippocampal region of the rat brain, without showing cytotoxicity. MATERIALS & METHODS: Fluorescence imaging with siRNA(Cy3)GAPDH and dark-field imaging using plasmonic enhanced scattering from GNRs were used to monitor the distribution of the nanoplexes within different neuronal cell types present in the targeted hippocampal region. RESULTS & CONCLUSION: Our results show robust nanoplex uptake and slow release of the fluorescent gene silencer with significant impact on the suppression of GAPDH gene expression (70% gene silencing, >10 days postinjection). The observed gene knockdown using nanoplexes in targeted regions of the brain opens a new era of drug treatment for neurological disorders.


Subject(s)
Gene Knockdown Techniques/methods , Gold/chemistry , Hippocampus/metabolism , Nanotubes/chemistry , Animals , Ganglia, Spinal/cytology , Glyceraldehyde 3-Phosphate Dehydrogenase (NADP+)/genetics , Glyceraldehyde 3-Phosphate Dehydrogenase (NADP+)/metabolism , Male , Nanotechnology , RNA, Small Interfering/chemistry , Rats , Rats, Sprague-Dawley
15.
Clin J Pain ; 25(2): 101-6, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19333153

ABSTRACT

BACKGROUND: Alpha2 adrenergic agonists have long been employed as analgesics and to sedate patients undergoing surgical procedures. In addition, their therapeutic response synergizes that elicited by opioids. Although this response is well known, the role of alpha2 agonists, such as clonidine, during various painful surgical procedures remains to be elucidated. The goal of our study was to evaluate the effects of the intrathecal administration of clonidine on postoperative pain control and time to extubation in patients undergoing coronary artery bypass grafting. METHODS: Eighty-five patients undergoing coronary artery bypass grafting randomly received either an intrathecal injection of preservative free morphine 0.5 mg (MOR) or a combination of morphine 0.5 mg and clonidine 100 microg (CMC) before induction of anesthesia. Anesthesia was induced and maintained using a balanced anesthesia technique. Patients were transferred to the intensive care unit while intubated and weaned from mechanical ventilation following an established weaning protocol. Postoperative pain, opioid use within the first 24 hours, and time to extubation were used as primary outcome variables. Data were analyzed by a 2-tailed t test for continuous variables and Fisher exact test for nonparametric variables. RESULTS: There were no demographic differences between the CMC and MOR groups. Postoperative pain, as assessed by a visual analog scale, was milder in the CMC group when compared with that of the MOR group (2.2+/-0.36 vs. 3.4+/-0.33, P<0.05). Similarly, patients in the CMC group required lower doses of morphine within 24 hours compared with the MOR group (2.02+/-0.36 vs. 6.47+/-0.49 mg, P<0.0001). Time to extubation was significantly shorter in patients receiving CMC than in those who received MOR (592+/-52 vs. 887+/-75 min, P<0.05). There was no mortality in either group. There was a trend for increased vasopressin use in the CMC group compared with the MOR group, although this was not statistically significant (P=0.07). CONCLUSIONS: Addition of clonidine to neuraxial opioids improves the quality of analgesia postoperatively and expedites the process of weaning from mechanical ventilation. There were no serious adverse events in the cohort of the patients studied. However, the safety profile of this medication remains to be examined with a larger group of patients.


Subject(s)
Analgesics/therapeutic use , Clonidine/therapeutic use , Coronary Artery Bypass/adverse effects , Pain, Postoperative/drug therapy , Pain, Postoperative/etiology , Aged , Chemotherapy, Adjuvant/methods , Female , Hemodynamics/drug effects , Humans , Injections, Spinal/methods , Male , Middle Aged , Morphine/therapeutic use , Narcotics/therapeutic use , Pain Measurement , Reaction Time/drug effects , Respiration/drug effects
16.
Eur J Pharmacol ; 588(2-3): 217-31, 2008 Jul 07.
Article in English | MEDLINE | ID: mdl-18514187

ABSTRACT

Tumor necrosis factor-alpha (TNF) plays a role in neuropathic pain. During neuropathic pain development in the chronic constriction injury model, elevated TNF levels in the brain occur in association with enhanced alpha 2-adrenoceptor inhibition of norepinephrine release. alpha 2-Adrenoceptors are also located on peripheral macrophage where they normally function as pro-inflammatory, since they increase the production of the cytokine TNF, a proximal mediator of inflammation. How the central increase in TNF affects peripheral alpha 2-adrenoceptor function was investigated. Male, Sprague-Dawley rats had four loose ligatures placed around the right sciatic nerve. Thermal hyperalgesia was determined by comparing hind paw withdrawal latencies between chronic constriction injury and sham-operated rats. Chronic constriction injury increased TNF immunoreactivity at the lesion and the hippocampus. Amitriptyline, an antidepressant that is used as an analgesic, was intraperitoneally administered (10 mg/kg) starting simultaneous with ligature placement (day-0) or at days-4 or -6 post-surgery. Amitriptyline treatment initiated at day-0 or day-4 post-ligature placement alleviated hyperalgesia. When initiated at day-0, amitriptyline prevented increased TNF immunoreactivity in the hippocampus and at the lesion. A peripheral inflammatory response, macrophage production of TNF, was also assessed in the current study. Lipopolysaccharide (LPS)-stimulated production of TNF by whole blood cells and peritoneal macrophages was determined following activation of the alpha 2-adrenoceptor in vitro. alpha 2-Adrenoceptor regulation of TNF production from peripheral immune-effector cells reversed from potentiation in controls to inhibition in chronic constriction injured rats. This effect is accelerated with amitriptyline treatment initiated at day-0 or day-4 post-ligature placement. Amitriptyline treatment initiated day-6 post-ligature placement did not alleviate hyperalgesia and prevented the switch from potentiation to inhibition in alpha 2-adrenoceptor regulation of TNF production. Recombinant rat TNF i.c.v. microinfusion reproduces the response of peripheral macrophages from rats with chronic constriction injury. A reversal in peripheral alpha 2-adrenoceptor regulation of TNF production from pro- to anti-inflammatory is associated with effective alleviation of thermal hyperalgesia. Thus, alpha 2-adrenoceptor regulation of peripheral TNF production may serve as a potential biomarker to evaluate therapeutic regimens.


Subject(s)
Analgesics/pharmacology , Inflammation/metabolism , Macrophages/metabolism , Monocytes/metabolism , Receptors, Adrenergic, alpha-2/physiology , Tumor Necrosis Factor-alpha/biosynthesis , Amitriptyline/therapeutic use , Animals , Lipopolysaccharides/pharmacology , Male , Rats , Rats, Sprague-Dawley , Sciatic Nerve/drug effects , Sciatic Nerve/pathology , Tumor Necrosis Factor-alpha/analysis
17.
Neuropharmacology ; 52(2): 576-89, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17055005

ABSTRACT

The central component that establishes chronic pain from peripheral nerve injury is associated with increased tumor necrosis factor-alpha (TNFalpha) production in the brain. This study examined TNFalpha and its reciprocally permissive role with alpha(2)-adrenergic activation during peak and progressive decline of thermal hyperalgesia in sciatic nerve chronic constriction injury (CCI). Accumulation of TNFalpha mRNA (in situ hybridization) increases in the hippocampus and locus coeruleus during the onset of neuropathic pain and persists as hyperalgesia abates. Activation of alpha(2)-adrenergic receptors in control rats decreases TNFalpha mRNA accumulation in these brain regions. In contrast, during hyperalgesia, alpha(2)-adrenergic activation enhances TNFalpha mRNA accumulation. Whether this enhanced TNFalpha production is associated with changes in the regulation of norepinephrine (NE) release was tested. Hippocampal slices were electrically depolarized to evaluate alpha(2)-adrenergic and TNFalpha regulation of NE release. While inhibition of NE release by TNFalpha is maximal during peak hyperalgesia, it subsequently transforms to facilitate NE release. In addition, alpha(2)-adrenergic receptor activation with clonidine (0.2mg/kg, i.p.) in CCI rats experiencing hyperalgesia restores TNFalpha and alpha(2)-adrenergic inhibition of NE release. While TNFalpha directs the development of hyperalgesia, it also directs its resolution. Transformed sensitivity to alpha(2)-adrenergic agonists during hyperalgesia demonstrates a mechanism for therapy.


Subject(s)
Hyperalgesia/metabolism , Norepinephrine/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adrenergic alpha-Agonists/pharmacology , Analysis of Variance , Animals , Brimonidine Tartrate , Clonidine/pharmacology , Clonidine/therapeutic use , Dose-Response Relationship, Radiation , Electric Stimulation/methods , Functional Laterality/drug effects , Functional Laterality/physiology , Hippocampus/drug effects , Hippocampus/metabolism , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Hyperalgesia/pathology , In Situ Hybridization/methods , In Vitro Techniques , Locus Coeruleus/drug effects , Locus Coeruleus/metabolism , Male , Pain Measurement , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Sciatica/complications , Sciatica/drug therapy , Sciatica/etiology
18.
Brain Behav Immun ; 21(1): 112-24, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16859892

ABSTRACT

Integral to neuropathic pain is a reciprocal interaction between tumor necrosis factor-alpha (TNF) production and the alpha(2)-adrenergic receptor response, offering an attractive therapeutic target. The effects of varying levels of brain TNF on alpha(2)-adrenergic regulation of cyclic AMP (cAMP) production in the hippocampus and sciatic nerve were investigated during the development and amitriptyline treatment of chronic pain. Increased levels of TNF during the development of chronic pain transform alpha(2)-adrenergic inhibition of cAMP production in the brain to potentiation. While alpha(2)-adrenergic receptors regulate TNF production, they also affect descending noradrenergic pathways. Increases in levels of TNF in the brain deeply impact peripheral inflammation through regulating alpha(2)-adrenergic receptors, offering insight into brain-body interactions during neuropathic pain. Amitriptyline as an analgesic inhibits pain-induced increases in brain-associated TNF and transforms peripheral alpha(2)-adrenergic receptors. The dynamic equilibrium between TNF levels and alpha(2)-adrenergic functioning is uniquely altered during development and treatment of neuropathic pain. Proper manipulations of this interaction offer efficacious treatment of neuropathic pain.


Subject(s)
Hippocampus/metabolism , Neuralgia/metabolism , Receptors, Adrenergic, alpha/metabolism , Sciatic Nerve/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adrenergic Uptake Inhibitors/pharmacology , Amitriptyline/pharmacology , Animals , Cyclic AMP/metabolism , Male , Nerve Crush , Neuralgia/psychology , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha/drug effects , Sciatic Nerve/drug effects , Sciatic Nerve/injuries , Signal Transduction/drug effects , Signal Transduction/physiology
19.
Neuropharmacology ; 48(3): 448-60, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15721177

ABSTRACT

Neuropathic pain, a chronic disabling pain arising from nerve injury, develops a central component. In brain neurons, tumor necrosis factor-alpha (TNF) levels intensify and TNF-inhibition of norepinephrine (NE) release, dependent upon alpha(2)-adrenergic activation, amplifies during neuropathic pain onset. TNF-inhibition of NE release transforms to facilitation in the hippocampus of rats administered antidepressants (treat neuropathic pain), contemporaneous with decreased neuron TNF. Therefore, adrenergic drugs inhibit increased pain sensitivity (hyperalgesia) by decreasing TNF production, thereby inducing increased NE release. This study examined TNF- and alpha(2)-adrenergic-regulated NE release from hippocampal slices during both the onset and dissipation of hyperalgesia during sciatic nerve chronic constriction injury (CCI). The enhanced inhibition of NE release by TNF at peak hyperalgesia (day-8) transformed to facilitation of NE release at days 12, 14, 16, and 21 post-CCI, corresponding to dissipation of hyperalgesia. Chronic antidepressant drug administration alone to rats results in similar findings. Rats administered the antidepressant amitriptyline (10 mg/kg, i.p., 60 min) at day-8 post-CCI, no longer exhibited hyperalgesia. Interestingly, the presynaptic response to TNF transformed to facilitation of NE release. While TNF directs the development of hyperalgesia, it is also involved in the resolution of pain, a possible mechanism for management of chronic pain.


Subject(s)
Pain Measurement/drug effects , Pain/drug therapy , Sciatic Neuropathy/drug therapy , Tumor Necrosis Factor-alpha/physiology , Animals , Dose-Response Relationship, Drug , Male , Norepinephrine/metabolism , Pain/metabolism , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Sciatic Neuropathy/metabolism , Tumor Necrosis Factor-alpha/therapeutic use
20.
J Neurosci Res ; 79(6): 779-87, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15672410

ABSTRACT

Alpha2-adrenergic receptors control norepinephrine (NE) release and tumor necrosis factor-alpha (TNF) production from neurons. TNF regulates NE release, depending on alpha2-adrenergic receptor functioning. The relationship between TNF production in the brain and alpha2-adrenergic receptor activation could have profound control over NE release. TNF and alpha2-adrenergic regulation of NE release was investigated in rat hippocampal slices incubated with pertussis toxin (PTX). The alpha2-adrenergic receptor couples to Galpha(i/o)-proteins to inhibit NE release; however, in slices preexposed to PTX, alpha2-adrenergic receptor activation facilitates NE release. TNF exposure subsequent to PTX restores alpha2-adrenergic inhibition of NE release. PTX exposure of hippocampal slices prevents agonist-induced increases in Galpha(i/o) labeling with a GTP analog; after subsequent TNF exposure, agonist-induced increases in Galpha(i/o) labeling are restored. TNF regulation of NE release transforms from inhibition to facilitation depending on alpha2-adrenergic receptor activation following PTX exposure. Therefore, TNF directs the coupling of the alpha2-adrenergic receptor, ultimately affecting NE release.


Subject(s)
GTP-Binding Proteins/physiology , Guanosine Triphosphate/analogs & derivatives , Hippocampus/drug effects , Norepinephrine/metabolism , Receptors, Adrenergic, alpha-2/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Animals , Azides/pharmacology , Brimonidine Tartrate , Cell Membrane/drug effects , Cell Membrane/radiation effects , Dose-Response Relationship, Drug , Drug Interactions , Electrophoresis, Polyacrylamide Gel/methods , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Guanosine Triphosphate/pharmacology , Hippocampus/metabolism , In Vitro Techniques , Male , Pertussis Toxin/pharmacology , Phenoxybenzamine/pharmacology , Phosphorus Isotopes/pharmacology , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-2/physiology , Yohimbine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL